Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Brain Behav Immun ; 111: 395-411, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37169133

RESUMEN

Aging coincides with major changes in brain immunity that aid in a decline in neuronal function. Here, we postulate that systemic, pro-aging factors contribute to immunological changes that occur within the brain during aging. To investigate this hypothesis, we comprehensively characterized the central and peripheral immune landscape of 20-month-old male mice using cytometry by time-of-flight (CyTOF) and investigated the role of age-associated circulating factors. We found that CD8+ T cells expressing programmed cell death protein 1 (PD1) and tissue-resident memory CD8+ T cells accumulated in the aged brain while levels of memory T cells rose in the periphery. Injections of plasma derived from 20-month-old mice into 5-month-old receiving mice decreased the frequency of splenic and circulating naïve T cells, increased memory CD8+ T cells, and non-classical, patrolling monocytes in the spleen, and elevated levels of regulatory T cells and non-classical monocytes in the blood. Notably, CD8+ T cells accumulated within white matter areas of plasma-treated mice, which coincided with the expression of vascular cell adhesion molecule 1 (VCAM-1), a mediator of immune cell trafficking, on the brain vasculature. Taken together, we here describe age-related immune cell changes in the mouse brain and circulation and show that age-associated systemic factors induce the expansion of CD8+ T cells in the aged brain.


Asunto(s)
Linfocitos T CD8-positivos , Linfocitos T Reguladores , Ratones , Masculino , Animales , Factores de Edad , Envejecimiento , Encéfalo
2.
Mol Psychiatry ; 25(7): 1382-1405, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31222184

RESUMEN

A decrease in adult hippocampal neurogenesis has been linked to age-related cognitive impairment. However, the mechanisms involved in this age-related reduction remain elusive. Glucocorticoid hormones (GC) are important regulators of neural stem/precursor cells (NSPC) proliferation. GC are released from the adrenal glands in ultradian secretory pulses that generate characteristic circadian oscillations. Here, we investigated the hypothesis that GC oscillations prevent NSPC activation and preserve a quiescent NSPC pool in the aging hippocampus. We found that hippocampal NSPC populations lacking expression of the glucocorticoid receptor (GR) decayed exponentially with age, while GR-positive populations decayed linearly and predominated in the hippocampus from middle age onwards. Importantly, GC oscillations controlled NSPC activation and GR knockdown reactivated NSPC proliferation in aged mice. When modeled in primary hippocampal NSPC cultures, GC oscillations control cell cycle progression and induce specific genome-wide DNA methylation profiles. GC oscillations induced lasting changes in the methylation state of a group of gene promoters associated with cell cycle regulation and the canonical Wnt signaling pathway. Finally, in a mouse model of accelerated aging, we show that disruption of GC oscillations induces lasting changes in dendritic complexity, spine numbers and morphology of newborn granule neurons. Together, these results indicate that GC oscillations preserve a population of GR-expressing NSPC during aging, preventing their activation possibly by epigenetic programming through methylation of specific gene promoters. Our observations suggest a novel mechanism mediated by GC that controls NSPC proliferation and preserves a dormant NSPC pool, possibly contributing to a neuroplasticity reserve in the aging brain.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Ritmo Circadiano , Glucocorticoides/metabolismo , Hipocampo/citología , Células-Madre Neurales/metabolismo , Animales , Encéfalo/citología , Proliferación Celular , Masculino , Ratones , Neurogénesis , Receptores de Glucocorticoides/metabolismo
3.
Neuropathol Appl Neurobiol ; 46(2): 142-159, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31183875

RESUMEN

AIM: Matrix metalloproteinases (MMPs) and their endogenous tissue inhibitors (TIMPs) control proteolysis within the extracellular matrix (ECM) of the brain. Dysfunction of this enzymatic system due to brain inflammation can disrupt the blood-brain barrier (BBB) and has been implicated in the pathogenesis of epilepsy. However, this has not been extensively studied in the epileptogenic human brain. METHODS: We investigated the expression and cellular localization of major MMPs (MMP2, MMP3, MMP9 and MMP14) and TIMPs (TIMP1, TIMP2, TIMP3 and TIMP4) using quantitative real-time polymerase chain reaction (RT-PCR) and immunohistochemistry in resected epileptogenic brain tissue from patients with tuberous sclerosis complex (TSC), a severe neurodevelopmental disorder characterized by intractable epilepsy and prominent neuroinflammation. Furthermore, we determined whether anti-inflammatory microRNAs, miR146a and miR147b, which can regulate gene expression at the transcriptional level, could attenuate dysregulated MMP and TIMP expression in TSC tuber-derived astroglial cultures. RESULTS: We demonstrated higher mRNA and protein expression of MMPs and TIMPs in TSC tubers compared to control and perituberal brain tissue, particularly in dysmorphic neurons and giant cells, as well as in reactive astrocytes, which was associated with BBB dysfunction. More importantly, IL-1ß-induced dysregulation of MMP3, TIMP2, TIMP3 and TIMP4 could be rescued by miR146a and miR147b in tuber-derived TSC cultures. CONCLUSIONS: This study provides evidence of dysregulation of the MMP/TIMP proteolytic system in TSC, which is associated with BBB dysfunction. As dysregulated MMP and TIMP expression can be ameliorated in vitro by miR146a and miR147b, these miRNAs deserve further investigation as a novel therapeutic approach.


Asunto(s)
Metaloproteinasas de la Matriz/metabolismo , MicroARNs/metabolismo , Esclerosis Tuberosa/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Preescolar , Humanos , Masculino , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Esclerosis Tuberosa/patología , Células Tumorales Cultivadas
4.
Neuropathol Appl Neurobiol ; 44(4): 404-416, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28543098

RESUMEN

AIMS: Cell matrix modulating protein SPARCL-1 is highly expressed by astrocytes during CNS development and following acute CNS damage. Applying NanoLC-MS/MS to CSF of RRMS and SPMS patients, we identified SPARCL-1 as differentially expressed between these two stages of MS, suggesting a potential as CSF biomarker to differentiate RRMS from SPMS and a role in MS pathogenesis. METHODS: This study examines the potential of SPARCL-1 as CSF biomarker discriminating RRMS from SPMS in three independent cohorts (n = 249), analyses its expression pattern in MS lesions (n = 26), and studies its regulation in cultured human brain microvasculature endothelial cells (BEC) after exposure to MS-relevant inflammatory mediators. RESULTS: SPARCL-1 expression in CSF was significantly higher in SPMS compared to RRMS in a Dutch cohort of 76 patients. This finding was not replicated in 2 additional cohorts of MS patients from Sweden (n = 81) and Switzerland (n = 92). In chronic MS lesions, but not active lesions or NAWM, a vessel expression pattern of SPARCL-1 was observed in addition to the expression by astrocytes. EC were found to express SPARCL-1 in chronic MS lesions, and SPARCL-1 expression was regulated by MS-relevant inflammatory mediators in cultured human BEC. CONCLUSIONS: Conflicting results of SPARCL-1's differential expression in CSF of three independent cohorts of RRMS and SPMS patients precludes its use as biomarker for disease progression. The expression of SPARCL-1 by BEC in chronic MS lesions together with its regulation by inflammatory mediators in vitro suggest a role for SPARCL-1 in MS neuropathology, possibly at the brain vascular level.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Esclerosis Múltiple/metabolismo , Adulto , Biomarcadores/metabolismo , Encéfalo/patología , Progresión de la Enfermedad , Células Endoteliales/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/patología
5.
Neurobiol Dis ; 107: 57-65, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27659108

RESUMEN

The blood-brain barrier (BBB) is indispensable for the maintenance of brain homeostasis and proper neuronal functioning. Dysfunction of the BBB significantly contributes to the pathogenesis of neuroinflammatory and neurodegenerative diseases like stroke, multiple sclerosis (MS), and Alzheimer's disease (AD). The neuroinflammatory environment that characterizes these disorders propagates chronic impaired function of the BBB, processes that will be discussed in this review. Limiting dysfunction of the BBB may be an attractive target for treatment of neurological disorders. To date, no current treatments are directly targeting the function of the BBB. In this review, we will specifically discuss the potential protective role of nuclear liver X receptors (LXRs) as a promising therapeutic target to reverse or prevent BBB impairment in neurological diseases.


Asunto(s)
Barrera Hematoencefálica/inmunología , Receptores X del Hígado/metabolismo , Animales , Humanos , Enfermedades Neurodegenerativas/metabolismo , Neuroinmunomodulación/fisiología , Neuroprotección/fisiología
6.
Drug Resist Updat ; 19: 1-12, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25791797

RESUMEN

Gliomas are the most common primary brain tumors. Particularly in adult patients, the vast majority of gliomas belongs to the heterogeneous group of diffuse gliomas, i.e. glial tumors characterized by diffuse infiltrative growth in the preexistent brain tissue. Unfortunately, glioblastoma, the most aggressive (WHO grade IV) diffuse glioma is also by far the most frequent one. After standard treatment, the 2-year overall survival of glioblastoma patients is approximately only 25%. Advanced knowledge in the molecular pathology underlying malignant transformation has offered new handles and better treatments for several cancer types. Unfortunately, glioblastoma multiforme (GBM) patients have not yet profited as although numerous experimental drugs have been tested in clinical trials, all failed miserably. This grim prognosis for GBM is at least partly due to the lack of successful drug delivery across the blood-brain tumor barrier (BBTB). The human brain comprises over 100 billion capillaries with a total length of 400 miles, a total surface area of 20 m(2) and a median inter-capillary distance of about 50 µm, making it the best perfused organ in the body. The BBTB encompasses existing and newly formed blood vessels that contribute to the delivery of nutrients and oxygen to the tumor and facilitate glioma cell migration to other parts of the brain. The high metabolic demands of high-grade glioma create hypoxic areas that trigger increased expression of VEGF and angiogenesis, leading to the formation of abnormal vessels and a dysfunctional BBTB. Even though the BBTB is considered 'leaky' in the core part of glioblastomas, in large parts of glioblastomas and, even more so, in lower grade diffuse gliomas the BBTB more closely resembles the intact blood-brain barrier (BBB) and prevents efficient passage of cancer therapeutics, including small molecules and antibodies. Thus, many drugs can still be blocked from reaching the many infiltrative glioblastoma cells that demonstrate 'within-organ-metastasis' away from the core part to brain areas displaying a more organized and less leaky BBTB. Hence, drug delivery in glioblastoma deserves explicit attention as otherwise new experimental therapies will continue to fail. In the current review we highlight different aspects of the BBTB in glioma patients and preclinical models and discuss the advantages and drawbacks of drug delivery approaches for the treatment of glioma patients. We provide an overview on methods to overcome the BBTB, including osmotic blood-brain barrier disruption (BBBD), bradykinin receptor-mediated BBTB opening, inhibition of multidrug efflux transporters, receptor-mediated transport systems and physiological circumvention of the BBTB. While our knowledge about the molecular biology of glioma cells is rapidly expanding and is, to some extent, already assisting us in the design of tumor-tailored therapeutics, we are still struggling to develop modalities to expose the entire tumor to such therapeutics at pharmacologically meaningful quantities. Therefore, we must expand our knowledge about the fundamentals of the BBTB as a step toward the design of practical and safe devices and approaches for enhanced drug delivery into the diseased brain area.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Adulto , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/patología , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Glioblastoma/patología , Humanos , Pronóstico
7.
Mult Scler ; 21(10): 1280-90, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25583833

RESUMEN

BACKGROUND: Cortical atrophy, assessed with magnetic resonance imaging (MRI), is an important outcome measure in multiple sclerosis (MS) studies. However, the underlying histopathology of cortical volume measures is unknown. OBJECTIVE: We investigated the histopathological substrate of MRI-measured cortical volume in MS using combined post-mortem imaging and histopathology. METHODS: MS brain donors underwent post-mortem whole-brain in-situ MRI imaging. After MRI, tissue blocks were systematically sampled from the superior and inferior frontal gyrus, anterior cingulate gyrus, inferior parietal lobule, and superior temporal gyrus. Histopathological markers included neuronal, axonal, synapse, astrocyte, dendrite, myelin, and oligodendrocyte densities. Matched cortical volumes from the aforementioned anatomical regions were measured on the MRI, and used as outcomes in a nested prediction model. RESULTS: Forty-five tissue blocks were sampled from 11 MS brain donors. Mean age at death was 68±12 years, post-mortem interval 4±1 hours, and disease duration 35±15 years. MRI-measured regional cortical volumes varied depending on anatomical region. Neuronal density, neuronal size, and axonal density were significant predictors of GM volume. CONCLUSIONS: In patients with long-standing disease, neuronal and axonal pathology are the predominant pathological substrates of MRI-measured cortical volume in chronic MS.


Asunto(s)
Atrofia/patología , Corteza Cerebral/patología , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Esclerosis Múltiple/patología , Lóbulo Parietal/patología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/diagnóstico , Enfermedades Neurodegenerativas/patología
8.
Front Aging ; 4: 1112109, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36911498

RESUMEN

Aging associates with an increased susceptibility for disease and decreased quality of life. To date, processes underlying aging are still not well understood, leading to limited interventions with unknown mechanisms to promote healthy aging. Previous research suggests that changes in the blood proteome are reflective of age-associated phenotypes such as frailty. Moreover, experimentally induced changes in the blood proteome composition can accelerate or decelerate underlying aging processes. The aim of this study is to identify a set of proteins in the human plasma associated with aging by integration of the data of four independent, large-scaled datasets using the aptamer-based SomaScan platform on the human aging plasma proteome. Using this approach, we identified a set of 273 plasma proteins significantly associated with aging (aging proteins, APs) across these cohorts consisting of healthy individuals and individuals with comorbidities and highlight their biological functions. We validated the age-associated effects in an independent study using a centenarian population, showing highly concordant effects. Our results suggest that APs are more associated to diseases than other plasma proteins. Plasma levels of APs can predict chronological age, and a reduced selection of 15 APs can still predict individuals' age accurately, highlighting their potential as biomarkers of aging processes. Furthermore, we show that individuals presenting accelerated or decelerated aging based on their plasma proteome, respectively have a more aged or younger systemic environment. These results provide novel insights in the understanding of the aging process and its underlying mechanisms and highlight potential modulators contributing to healthy aging.

9.
Cell Death Dis ; 14(11): 781, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-38016947

RESUMEN

In Alzheimer's disease (AD) more than 50% of the patients are affected by capillary cerebral amyloid-angiopathy (capCAA), which is characterized by localized hypoxia, neuro-inflammation and loss of blood-brain barrier (BBB) function. Moreover, AD patients with or without capCAA display increased vessel number, indicating a reactivation of the angiogenic program. The molecular mechanism(s) responsible for BBB dysfunction and angiogenesis in capCAA is still unclear, preventing a full understanding of disease pathophysiology. The Liver X receptor (LXR) family, consisting of LXRα and LXRß, was reported to inhibit angiogenesis and particularly LXRα was shown to secure BBB stability, suggesting a major role in vascular function. In this study, we unravel the regulatory mechanism exerted by LXRα to preserve BBB integrity in human brain endothelial cells (BECs) and investigate its role during pathological conditions. We report that LXRα ensures BECs identity via constitutive inhibition of the transcription factor SNAI2. Accordingly, deletion of brain endothelial LXRα is associated with impaired DLL4-NOTCH signalling, a critical signalling pathway involved in vessel sprouting. A similar response was observed when BECs were exposed to hypoxia, with concomitant LXRα decrease and SNAI2 increase. In support of our cell-based observations, we report a general increase in vascular SNAI2 in the occipital cortex of AD patients with and without capCAA. Importantly, SNAI2 strongly associated with vascular amyloid-beta deposition and angiopoietin-like 4, a marker for hypoxia. In hypoxic capCAA vessels, the expression of LXRα may decrease leading to an increased expression of SNAI2, and consequently BECs de-differentiation and sprouting. Our findings indicate that LXRα is essential for BECs identity, thereby securing BBB stability and preventing aberrant angiogenesis. These results uncover a novel molecular pathway essential for BBB identity and vascular homeostasis providing new insights on the vascular pathology affecting AD patients.


Asunto(s)
Enfermedad de Alzheimer , Angiopatía Amiloide Cerebral , Humanos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/metabolismo , Angiopatía Amiloide Cerebral/complicaciones , Angiopatía Amiloide Cerebral/metabolismo , Angiopatía Amiloide Cerebral/patología , Células Endoteliales/metabolismo , Hipoxia/metabolismo , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo
10.
Adv Drug Deliv Rev ; 188: 114398, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35780907

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia affecting millions of people worldwide. While different immunotherapies are imminent, currently only disease-modifying medications are available and a cure is lacking. Over the past decade, immunological interfaces of the central nervous system (CNS) and their role in neurodegenerative diseases received increasing attention. Specifically, emerging evidence shows that subsets of circulating CD8+ T cells cross the brain barriers and associate with AD pathology. To gain more insight into how the adaptive immune system is involved in disease pathogenesis, we here provide a comprehensive overview of the contribution of T cells to AD pathology, incorporating changes at the brain barriers. In addition, we review studies that provide translation of these findings by targeting T cells to combat AD pathology and cognitive decline. Importantly, these data show that immunological changes in AD are not confined to the CNS and that AD-associated systemic immune changes appear to affect brain homeostasis.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Linfocitos T CD8-positivos/patología , Humanos , Sistema Inmunológico/patología
11.
Fluids Barriers CNS ; 18(1): 46, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34666786

RESUMEN

With the introduction of endovascular thrombectomy (EVT), a new era for treatment of acute ischemic stroke (AIS) has arrived. However, despite the much larger recanalization rate as compared to thrombolysis alone, final outcome remains far from ideal. This raises the question if some of the previously tested neuroprotective drugs warrant re-evaluation, since these compounds were all tested in studies where large-vessel recanalization was rarely achieved in the acute phase. This review provides an overview of compounds tested in clinical AIS trials and gives insight into which of these drugs warrant a re-evaluation as an add-on therapy for AIS in the era of EVT. A literature search was performed using the search terms "ischemic stroke brain" in title/abstract, and additional filters. After exclusion of papers using pre-defined selection criteria, a total of 89 trials were eligible for review which reported on 56 unique compounds. Trial compounds were divided into 6 categories based on their perceived mode of action: systemic haemodynamics, excitotoxicity, neuro-inflammation, blood-brain barrier and vasogenic edema, oxidative and nitrosative stress, neurogenesis/-regeneration and -recovery. Main trial outcomes and safety issues are summarized and promising compounds for re-evaluation are highlighted. Looking at group effect, drugs intervening with oxidative and nitrosative stress and neurogenesis/-regeneration and -recovery appear to have a favourable safety profile and show the most promising results regarding efficacy. Finally, possible theories behind individual and group effects are discussed and recommendation for promising treatment strategies are described.


Asunto(s)
Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Humanos
12.
PLoS One ; 14(4): e0213069, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30947313

RESUMEN

Extracellular vesicles (EVs) released by cells have a role in intercellular communication to regulate a wide range of biological processes. Two types of EVs can be recognized. Exosomes, which are released from multi-vesicular bodies upon fusion with the plasma membrane, and ectosomes, which directly bud from the plasma membrane. How cells regulate the quantity of EV release is largely unknown. One of the initiating events in vesicle biogenesis is the regulated transport of phospholipids from the exoplasmic to the cytosolic leaflet of biological membranes. This process is catalyzed by P4-ATPases. The role of these phospholipid transporters in intracellular vesicle transport has been established in lower eukaryotes and is slowly emerging in mammalian cells. In Caenorhabditis elegans (C. elegans), deficiency of the P4-ATPase member TAT-5 resulted in enhanced EV shedding, indicating a role in the regulation of EV release. In this study, we investigated whether the mammalian ortholog of TAT-5, ATP9A, has a similar function in mammalian cells. We show that knockdown of ATP9A expression in human hepatoma cells resulted in a significant increase in EV release that was independent of caspase-3 activation. Pharmacological blocking of exosome release in ATP9A knockdown cells did significantly reduce the total number of EVs. Our data support a role for ATP9A in the regulation of exosome release from human cells.


Asunto(s)
Adenosina Trifosfatasas/genética , Exosomas/genética , Vesículas Extracelulares/genética , Proteínas de Transporte de Membrana/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Caspasa 3/genética , Comunicación Celular/genética , Membrana Celular/genética , Micropartículas Derivadas de Células/genética , Endocitosis/genética , Vesículas Extracelulares/metabolismo , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Fosfolípidos/metabolismo , Transporte de Proteínas/genética
13.
Mult Scler Relat Disord ; 28: 44-49, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30553168

RESUMEN

BACKGROUND: Multiple sclerosis (MS) lacks reliable biomarkers that reflect disease activity. Recent evidence suggests that an altered sphingolipid metabolism is associated with MS pathogenesis. OBJECTIVE: To explore acid sphingomyelinase (ASM) activity and altered sphingolipid metabolism as potential biomarkers in serum of MS patients, to predict active and progressive disease, and response to disease modifying therapy (DMT). METHODS: Levels of serum ASM activity were longitudinally analyzed in 40 clinically isolated syndrome, 64 relapsing remitting (RR) and 10 primary progressive MS patients, and 22 healthy controls (HC). ASM activity and sphingolipid levels were measured in a different sample of 61 RRMS patients using DMT. RESULTS: A significant difference in ASM activity levels was observed between MS patients and HC (p < 0.001). There was no correlation between ASM activity levels and disease activity, progression or response to DMT. Ceramide (Cer)-C16:0 , Cer-C24:0 and sphingomyelin (SM)-C20:0, SM-C22:0, SM-C24:0 and SM-C24:1 showed a significant increase during fingolimod use. CONCLUSION: Although higher levels in MS patients were found, ASM activity levels do not show potential as a biomarker for predicting disease activity, progression or response to DMT. Two ceramides and four types of sphingomyelin require further investigation as potential markers for treatment response.


Asunto(s)
Enfermedades Desmielinizantes/sangre , Enfermedades Desmielinizantes/enzimología , Esfingomielina Fosfodiesterasa/sangre , Adulto , Biomarcadores/sangre , Ceramidas/sangre , Enfermedades Desmielinizantes/diagnóstico por imagen , Enfermedades Desmielinizantes/terapia , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Estudios de Seguimiento , Humanos , Factores Inmunológicos/uso terapéutico , Estudios Longitudinales , Masculino , Estudios Prospectivos , Esfingomielinas/sangre , Resultado del Tratamiento
14.
Free Radic Biol Med ; 45(12): 1729-37, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18930811

RESUMEN

Reactive oxygen species (ROS) and subsequent oxidative damage may contribute to the formation and persistence of multiple sclerosis (MS) lesions by acting on distinct pathological processes. ROS initiate lesion formation by inducing blood-brain barrier disruption, enhance leukocyte migration and myelin phagocytosis, and contribute to lesion persistence by mediating cellular damage to essential biological macromolecules of vulnerable CNS cells. Relatively little is known about which CNS cell types are affected by oxidative injury in MS lesions. Here, we show the presence of extensive oxidative damage to proteins, lipids, and nucleotides occurring in active demyelinating MS lesions, predominantly in reactive astrocytes and myelin-laden macrophages. Oxidative stress can be counteracted by endogenous antioxidant enzymes that confer protection against oxidative damage. Here, we show that antioxidant enzymes, including superoxide dismutase 1 and 2, catalase, and heme oxygenase 1, are markedly upregulated in active demyelinating MS lesions compared to normal-appearing white matter and white matter tissue from nonneurological control brains. Particularly, hypertrophic astrocytes and myelin-laden macrophages expressed an array of antioxidant enzymes. Enhanced antioxidant enzyme production in inflammatory MS lesions may reflect an adaptive defense mechanism to reduce ROS-induced cellular damage.


Asunto(s)
Antioxidantes/metabolismo , Catalasa/metabolismo , Hemo-Oxigenasa 1/metabolismo , Esclerosis Múltiple/enzimología , Esclerosis Múltiple/patología , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Astrocitos/enzimología , Encéfalo/enzimología , Encéfalo/patología , Estudios de Casos y Controles , Femenino , Humanos , Técnicas para Inmunoenzimas , Macrófagos/enzimología , Masculino , Persona de Mediana Edad , Superóxido Dismutasa-1
15.
Vascul Pharmacol ; 89: 12-18, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27894893

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia, affecting millions of people worldwide. One of the prominent causative factors of AD pathogenesis is cerebral vascular dysfunction, which results in diminished cerebral perfusion. Moreover, due to the loss of the protective function of the blood-brain barrier (BBB), impaired clearance of excess neurotoxic amyloid beta (Aß) occurs, causing vascular perturbation and diminished cognitive functioning. The relationship between the prevalence of AD and vascular risk factors is complex and not fully understood. In this review we illustrate the vascular risk factors, their effects on BBB function and their contributions to the onset of AD. Additionally, we discuss the underlying factors that may lead to altered neurovascular function and/or cerebral hypoperfusion in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Proteínas Angiogénicas/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Circulación Cerebrovascular , Cognición , Progresión de la Enfermedad , Humanos , Neovascularización Patológica , Prevalencia , Pronóstico , Medición de Riesgo , Factores de Riesgo
16.
J Neuroimmunol ; 52(1): 1-8, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7911476

RESUMEN

The presence and upregulation of adhesion molecules on bovine brain endothelial cells (BBEC) were investigated. Monolayers of BBEC were incubated with lipopolysaccharide (LPS), interleukin-1 beta (rhIL-1 beta), and interleukin-6 (rhIL-6) to simulate in vitro an inflammatory site in the cerebral capillaries. Adhesion of lymphocytes to BBEC increased 4.1-fold after stimulation of the endothelial cells for 4 h with 5 or 10 ng/ml LPS. Lymphocyte adhesion increased after incubation of the BBEC for 4 h with IL-1 and was increased 3.7-fold using 100 ng/ml IL-1. BBEC pre-incubated with IL-6 for 4 h also showed an increase in adhesion of lymphocytes, and cells pretreated with 100 ng/ml IL-6 showed a 3-fold increase in lymphocyte adherence. Specific monoclonal antibodies directed against CD11a, CD18, and VLA-4 were able to block adherence of lymphocytes to stimulated BBEC. These results indicate that the in vitro activation of BBEC may serve as a model for the study of inflammation of the blood-brain barrier.


Asunto(s)
Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Endotelio Vascular/citología , Linfocitos/fisiología , Animales , Antígenos CD/fisiología , Antígenos CD18 , Capilares/citología , Bovinos , Adhesión Celular , Moléculas de Adhesión Celular/fisiología , Células Cultivadas , Endotelio Vascular/fisiología , Molécula 1 de Adhesión Intercelular , Lipopolisacáridos/farmacología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Receptores de Antígeno muy Tardío/fisiología
17.
J Neuroimmunol ; 64(1): 37-43, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8598388

RESUMEN

The effects of the cytokines tumour necrosis factor-alpha (TNF-alpha), interleukin (IL)-1 beta and IL-6 on the permeability of monolayers of rat cerebral endothelial cells (RCEC) were investigated to assess potential changes in the integrity of the blood-brain barrier (BBB). RCEC were cultured to tight monolayers with a trans endothelial electrical resistance (TEER) of 100-150 ohm . cm2 on polycarbonate filters. Exposure of the RCEC to TNF-alpha, IL-1 beta and IL-6 induced a decline in the TEER, which could be completely abolished by 1 muM of indomethacin, a cyclooxygenase inhibitor. In addition, the effect of IL-1 beta on TEER across monolayers of RCEC could be completely inhibited by IL-1 receptor antagonist. In conclusion, cytokines induce a disruption of the BBB in vitro. In this process, cyclooxygenase activation within the endothelial cells seems to play a key role.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Citocinas/farmacología , Animales , Células Cultivadas , Interleucina-1/farmacología , Interleucina-6/farmacología , Prostaglandina-Endoperóxido Sintasas/fisiología , Ratas , Factor de Necrosis Tumoral alfa/farmacología
18.
J Neuroimmunol ; 59(1-2): 1-8, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7797610

RESUMEN

The capacity of rat cerebral endothelial cells (RCEC) to form eicosanoids was determined after incubation with 14C-labelled arachidonic acid. Prostaglandin E2 (PGE2) was the main metabolite formed by RCEC and was responsible for 54% of the total amount of eicosanoids produced. In contrast, in primary cultures of rat aorta endothelial cells, 32% of the amount of prostaglandins was 6-keto-PGF1 alpha). RCEC treated with 50 ng/ml LPS for 24 h responded with an augmented PGE2 synthesis and 6-keto-PGF1 alpha of 3.4-fold and 2.2-fold, respectively. Cultures treated with IL-1 beta (50 ng/ml) for 3 h showed a stimulation of the release of PGE2 and 6-keto-PGF1 alpha of 2.5- and 4.5-fold, respectively, and 2.0-fold and 2.3-fold, respectively, after IL-6 (50 ng/ml) incubation for 3 h. PGE2 is the main eicosanoid formed by RCEC in response to inflammatory agents, suggesting an important role of the cerebral endothelial cells in the transduction of an inflammatory response in the central nervous system.


Asunto(s)
Encéfalo/metabolismo , Eicosanoides/biosíntesis , Endotelio Vascular/metabolismo , Interleucina-1/farmacología , Interleucina-6/farmacología , Lipopolisacáridos/farmacología , Animales , Barrera Hematoencefálica , Células Cultivadas , Ratas , Ratas Wistar
19.
Biochem Pharmacol ; 50(2): 271-3, 1995 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-7632172

RESUMEN

Primary cultures of bovine cerebral endothelial cells were used as an in vitro model for the blood-brain barrier to study the transport and interactions of high-density lipoprotein (HDL) across monolayers of these cells. Transport of 125I-apoE free HDL across a monolayer of bovine cerebral endothelial cells occurred in a linear fashion up to a concentration of 70 micrograms/mL, suggesting paracellular transport of HDL. Bovine cerebral endothelial cells possess a high affinity binding site for HDL with a mean dissociation constant (KD) of 10.8 +/- 2.6 micrograms/mL (N = 4). Maximal binding of apoE free HDL to cerebral endothelial cells proved to be temperature-dependent: at 4 degrees a Bmax value of 42 +/- 9.3 ng/mg cell protein was found, while at 37 degrees this value was 177 +/- 70.4 ng/mg cell protein. Cell association of 125I-HDL could be effectively displaced by HDL, not by low-density lipoprotein or acetylated low-density lipoprotein, and association was not coupled to degradation. The in vitro blood-brain barrier cell system possesses high affinity binding sites for HDL, which are probably not involved in the transport of HDL across cerebral endothelial cells.


Asunto(s)
Encéfalo/metabolismo , Lipoproteínas HDL/metabolismo , Animales , Apolipoproteínas E/metabolismo , Transporte Biológico , Barrera Hematoencefálica , Bovinos , Células Cultivadas , Endotelio/metabolismo , Radioisótopos de Yodo , Temperatura
20.
Hum Pathol ; 20(11): 1071-7, 1989 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-2530150

RESUMEN

Four human fetuses with the limb body wall malformation complex, also known as the amniotic band syndrome, were examined. Besides malformations of the body wall and the limb(s), anomalies of the internal organs were found in three fetuses, suggesting a disturbance of normal morphogenesis before the fifth week of development. When analyzing the observed data in relation to their etiology, no support was found for either the amniotic band or the vascular disruption theory. New insights into morphogenesis suggest that the limb body wall malformation complex results from a malfunction in the ectodermal placodes and that it can be considered to be an embryonic dysplasia.


Asunto(s)
Músculos Abdominales/anomalías , Anomalías Múltiples/etiología , Deformidades Congénitas de las Extremidades , Tórax/anomalías , Músculos Abdominales/embriología , Adulto , Ectodermo/fisiología , Extremidades/embriología , Femenino , Humanos , Modelos Biológicos , Morfogénesis , Embarazo , Tórax/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA